The association between genomic variations and histological grade in hepatocellular carcinoma
Original Article

The association between genomic variations and histological grade in hepatocellular carcinoma

Jun Liu1, Guangbing Li1, Yuan Guo2, Ning Fan2, Yunjin Zang2

1Department of Hepatobiliary Surgery and Liver Transplantation, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China; 2Organ Transplant Center, the Affiliated Hospital of Qingdao University, Qingdao 266000, China

Contributions: (I) Conception and design: Y Zang; (II) Administrative support: Y Zang; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: J Liu; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Yunjin Zang. Organ Transplant Center, the Affiliated Hospital of Qingdao University, Qingdao 266000, China. Email: zangyj3657@qq.com.

Background: Histological grade (HG) is an important prognostic factor for hepatocellular carcinoma. With the development of precision medicine, diagnosis with a sequencing technology has become increasingly accepted. It is vital to discuss their similarities and differences to bridge or improve the traditional HG diagnosis with the novel sequencing technique.

Methods: A total of 658 tumor samples were collected from 602 Chinese hepatocellular carcinoma patients and sequenced for a panel of pan-cancer genes. Nucleotide usage bias, genomic variation-related scores, driver genes, and biological processes were compared among different HGs. These results were further verified using a cohort dataset from the Western population.

Results: Genomic variation subtypes, such as C>G substitution, maximum somatic allele frequency (MSAF), and TP53, and biological processes including “angiogenesis” and “regulation of homotypic cell-cell adhesion” were found to be significantly associated with HG in both Chinese and Western populations.

Conclusions: The association identified between genomic variation and HG could aid our understanding of HG as an important clinical measure, and potentially be used to predict HG for hepatocellular carcinoma.

Keywords: Histological grade (HG); hepatocellular carcinoma; sequencing; genomic variation


Submitted Oct 29, 2019. Accepted for publication Feb 17, 2020.

doi: 10.21037/tcr.2020.03.32


Introduction

Histological grade (HG) describes the aggressive potential of solid tumors. The classical and widely adopted grading system for hepatocellular carcinoma is Edmondson-Steiner (ES), which is based on microscopic evaluation of the tubule formation, mitotic count, and nuclear pleomorphism. According to the ES grading system, tumors can be classified into three or four grades. A tumor of a higher grade tends to grow and spread at a faster pace, which needs more urgent and aggressive treatment.

Needle biopsies and histopathological evaluation works as a gold standard for HG diagnosis. Collectively, clinical physicians have accumulated a large amount of experience in using this method. However, it has two major problems, diagnostic subjectivity and biopsy inaccessibility (1), which might hinder its full efficacy. A stricter tumor grading requires two or more pathologists with expertise in a specific cancer to reduce diagnostic subjectivity. Much effort has been made with non-invasive methods such as magnetic resonance and contrast computed tomography (CT) to avoid biopsy unavailability (2). In contrast to diverse imaging methods, molecular biomarkers could overcome the two problems mentioned above. For example, miR-1290 could work as a biomarker of high-grade serous ovarian carcinoma (3), and tumor tissue protein signatures could predict the HG of breast cancer (4). Apart from the expression of biomarkers as an indicator of HG, the genomic variation could also be used. Many gene mutations have been recognized to be associated with HG, such as TP53 (5), IDH1/2 (6) and ACVR2 (7). We are interested to know how far genomic variations are associated with HG in HCC because it can help us to understand HG as an important clinical measure.

With the development of precision medicine, DNA sequencing provides rich information for disease diagnosis and precision treatment. By using a liquid biopsy, genomic variations could prove to be more useful in predicting HG and could perfectly overcome the two major problems in the traditional ES grading system. Additionally, this method could provide necessary information for precise treatment in one-shot sequencing. However, ctDNA concentration is more easily affected by cancer development and clinical therapy, and the standard to detect ctDNA from liquid biopsy has not been well established.

This study sequenced 487 tissue samples from 459 Chinese HCC patients to build a solid connection between genomic variation and HG. Genomic variation, including nucleotide substitution/indel, truncation, gene homozygous deletion and fusion, were called. Association of HG with factors including genomic variation types, substitution types, mutational frequency related scores and biological processes was studied. Among the factors, those found to be significant were compared to those of the Western population.


Methods

Patients and samples

This study was approved by Shandong Provincial Hospital Affiliated to Shandong University and The Affiliated Hospital of Qingdao University. A total of 602 patients were enrolled. Each participant provided written informed consent. Samples were collected from surgery after diagnosis or relapse. HG was scored by a specialist in hepatobiliary pathology according to the Edmondson and Steiner method (8). Grade 1 was defined as well differentiated (WD), grade 2 and 3 as moderate differentiated (MD), grade 4 as poor differentiated (PD). The patients were staged according to the seventh edition of the tumor-node-metastasis (TNM) classification system for lung cancer from the American Joint Committee. We also collected another public dataset to validate our analysis. This dataset, MSKCC, containing 360 samples, was downloaded from cBioportal (https://www.cbioportal.org/, accessed on March 5, 2019).

Library preparation and next-generation sequencing

Tissue samples (40 µm section) were collected for each patient. KAPA Hyper Prep Kit (#07962363001, Roche, Basel, Switzerland) was used to extract DNA. PBS (phosphate-buffered saline) was added to those samples with volumes of less than 5 mL in order to make each sample volume equivalent to 5 mL. They were centrifuged (2 times at 1,600 g for 10 and 15 min, respectively) for extraction of DNA and the supernatant was separated. Invitrogen Qubit® DNA HS Assay Kit (#Q32854) was used to measure the DNA concentration. Single strand DNA and protein contamination were excluded. Library construction was only applied in samples with at least 50 ng of double-stranded DNA extracted. Molecular identifiers (MIDs) were added to the DNA segment ends for DNA libraries to reduce the false discovery rate (FDR). Barcodes were also added to the reads for multiplex sequencing. Sequencing was performed on an Illumina Novaseq 6000 (Illumina, San Diego, CA) for 151 bp read length from both ends. The average sequencing depth was about 3,000×.

Variants calling

A pan-cancer panel (Yuansuo®, Origimed, Shanghai, China) comprising 588 genes was captured with targeted amplification. Adaptors were trimmed from raw DNA reads by cutadapt (version 1.18) (9). MID-labeled reads were de-duplicated with an in-house pipeline. BWA MEM (version 0.7.9a) (10) mapped the high-quality reads to the UCSC hg19 reference sequences. Base quality was recalibrated by the BaseRecalibrator tool from GATK (version 3.8) (11). Mutect2 with a tumor-only mode (12) and Varscan (version 2.3.9) (13) with the default parameters were used to call variants.

For each sample, the germline variants having variant allele frequency (VAF) <0.1% were filtered according to the databases of ExAC (14), gnomAD (15), 1000 Genomes (16), and ESP6500 (17). Somatic variants that had not been filtered were further annotated by ANNOVAR (2017/07/17) (18) with RefSeq (version 2017/06/01).

Fermi-lite (19) was used to identify gene fusion and rearrangement. The breakpoints were further checked by BLAT (http://genome.ucsc.edu, version 3.50). Those reads uniquely mapping to the reference genome constituted rearrangement supported reads.

CNVKit (20) was used to estimate the logR scores. Copy number was assigned 1 for logR values below −0.25, 3 for logR values above 0.25, and 2 for logR values in between −0.25 and 0.25 (21).

Bioinformatics analysis

The mutant allele tumor heterogeneity (MATH) score for a tumor was calculated as the median absolute deviation divided by the median MAF of all somatic mutations detected in the tumor sample. As suggested by Jiang et al. (22), the calculation of MATH used somatic mutation calls with MAF of 0.075 or greater. Clonal mutation burden (CMB) (22) was defined as the number of mutations per clone, and divided into low (low TMB, high MATH), high (high TMB, low MATH), or intermediate (others).

Statistical analysis

The Mann-Whitney U test was used to compare TMB, MSAF, MATH, and CMB between different HGs. Fisher’s exact test was performed to compare the count number of nucleotide mutations for different HGs.

Survival analysis was conducted with R software. Samples were classified by a cutoff at the median mutational frequency. The survival time was plotted against overall survival probability by the Kaplan-Meier method. The log-rank test was applied to calculate the P value between the two groups.


Results

Patients and genomic variation detection

The analysis workflow of this study is illustrated in Figure S1. Initially, a total of 602 patients were enrolled in this study. Of these, only 459 patients had histological grading information available. The other patients’ samples were thus filtered out from the following study. Their clinicopathologic characteristics are summarized in Table S1. The median age of patients was 55 years old (range, 16 to 82 years old). Most of the patients were male (87.6%). According to the TNM classification system (23), the number of patients in the early stage (I/II/III) and late-stage (IV) were 418 and 41, respectively. Patients who consumed alcohol more than 200 days per year were classified as “drinking”, and those who had an immediate family member with any type of cancer were labeled as “family history”. HGs were divided into three categories: poorly differentiated (PD), moderately differentiated (MD) and well differentiated (WD).

Tissue samples were prepared by surgery and enriched with a pan-cancer panel of genes (Yuansuo®, Origimed Co., Ltd, Shanghai, China) (Figure S2). For the 459 patients, 487 samples were collected. Somatic genomic variations (SGVs) were called by Mutec2 (12) and Varscan (13) for each sample. Gene amplifications were called by CNVKit (20). Fermi-lite (19) was used to identify gene fusion and rearrangement. The genomic variations at top high frequency are depicted in Figure 1. Genomic variations from multiple samples of each patient were merged under the same patient.

Figure 1 The landscape of genomic variations. From top to bottom, the bar plot indicates the tumor mutation burdens (TMBs) and the below heat map indicated the clinicopathological characteristics. HG (histological grades) includes WD (well-differentiated), MD (moderately differentiated), and PD (poorly differentiated). The bottom left bar plot indicates the percentage of genomic variation for each gene in the patients. The bottom right heatmap shows genomic variation types.

The bias of SGV types in different HGs

There are multiple types of SGVs deriving from different mechanisms. Those SGVs were classified into five types (fusion/rearrangement, gene amplification, gene homozygous deletion, substitution/indel, and truncation). The percentage of those groups was summarized according to the HG groups (Figure 2A). From poorly to moderately to well-differentiated HG, the percentage of the truncation and substitution/indel group increased but that of the gene amplification group dropped. The poorly differentiated group had the lowest percentage of fusion/rearrangement but the highest percentage of gene amplification variations.

Figure 2 Variation distribution for hepatocellular carcinoma (HCC). (A) The upper plot shows the distribution of five types of genomic variation for three groups of HGs (histological grades). The lower plot is the distribution of 12 substitution types, which are grouped into transition and transversion. The x-axis indicates the patient percentage and the y-axis indicated the HGs. (B) The upper and the lower plots show MSAF (maximum somatic allele frequency) and MATH (mutant allele tumor heterogeneity) distributions for the three HG groups, respectively.

Single nucleotide variants (SNVs) can be classified into transversion substitution and transition substitution. Transition SNVs regularly had a higher frequency and caused no functional change because of codon “wobble”. To study the association between amino acid changes and HGs, the percentages of transversion and transition were compared (Figure 2A). In total, the percentage range of transversion and transition for different HGs was 65–72% and 28–34%, respectively. WD had higher transition than PD with percentages of 52% and 47%, respectively (P value =2.537e-11), but had lower transversion than PD with percentages of 48% and 53%, respectively (P value =2.2e-16). For specific substitutions, WD had less C>G transversion (P value =0.0058) and more G>A transition (P value =0.026) than non-WD. PD had higher C>T transition (P value =0.01) than non-PD.

Except for mutational occurrence, we also studied the association between HGs and mutational frequency related scores including maximum somatic allele frequency (MSAF) (24), MATH and CMB (22). MSAF was regularly used as a measure of cellular tumor prevalence. Higher MSAF denoted higher tumor content. The MATH score denoted allele heterogeneity among each sample, which reflected the diversity of mutational clones. CMB score combined the tumor mutation burden (TMB) and MATH score (22). High CMB was defined as high TMB and low MATH. These scores were compared among different HGs. WD had significantly lower MSAF than MD and PD with P values equal to 0.031 and 0.038, respectively (Figure 2B). As for the MATH score, MD was highest among HGs, but only MD transversion. PD had a P value of less than 0.05. We also tested the CMB score, but no significant difference was found among HGs (result not shown).

The functional bias of genomic variations for different HGs

Driver genes play a big part in cancer. Their specific effect on HG was also studied. The driver genes of HCC were collected from the literature (25,26). The top 10 variable genes are displayed in Figure 3A. Genes TP53, TERT, CTNNB1, RB1, AXIN1, and ARID1A were prone to substitution/indel/truncation variation, while CCND1, FGF19, FGF4, and FGF3 preferred gene amplification. Among those driver genes, there were three genes showing significantly different HGs after mutation (Figure 3B). Of these three genes, mutational TP53 (TP53+) had a higher average HG than non-mutational TP53 (TP53). A non-parameter Wilcoxon’s rank-sum test showed significance at P value =3.8e-2. In contrast, mutational CTNNB1 (CTNNB1+) and FGF3 (FGF3+) showed significantly lower HG with P value =7.8e-4 and P value =0.04, respectively. We also tested the association between gene amplification variation and HG for those driver genes, but no significant difference was found (Figure 3C).

Figure 3 Frequency of genomic variation in driver genes. (A) The percentage of patients with five types of genomic variations for hepatocellular carcinoma (HCC) driver genes. (B) The average grade of tumors with/without substitution/indel/truncation mutations in driver genes. *, P<0.05; **, P<0.01; ***, P<0.001. (C) The average grade of tumors with/without amplification in driver genes. SNV, single-nucleotide variant; CNV, copy number variation.

Mutations, such as substitution, indel, and truncation, can modify the targeted gene functions, and amplification can modify their expression. To study their functional bias, three HG groups were intersected with each other as displayed by a Venn plot in Figure 4A. WD, MD, and PD had 14, 57 and 62 unique mutated genes, respectively. WD had fewer unique mutated genes than other HGs. The unique genes for WD and PD were enriched with the biological processes of gene ontology. A hypergeometric test was performed for each biological process. The Bonferroni-Hochberg (BH) method was applied to correct for multiple testing errors. The top 10 enriched biological processes are listed in Figure 4B,C,D. WD was enriched in the regulation of the developmental process, cell differentiation, and membrane invagination. There were 1,168 biological processes enriched for PD specific mutations with multiple testing corrected P values less than 0.05, such as cell proliferation, protein phosphorylation, and cellular response to a stimulus.

Figure 4 The substitution/indel/truncation overlaps between different histological grades. (A) The substitution/indel/truncation overlaps between three HGs including WD, MD, and PD; (B) the enriched biological processes for well-differentiated tumors; (C) the enriched biological processes for moderately differentiated tumors; (D) the enriched biological processes for poorly differentiated tumors. The length of the blue bar indicates the negative log-transformed false discovery rate (FDR). HGs, histological grades; WD, well-differentiated; MD, moderately differentiated; PD, poorly differentiated.

Apart from substitution/indel/truncation mutations, copy number variation can also disrupt cellular function by modifying gene regulation. The amplified genes were intersected with each other (Figure S3A) to obtain the HG-specific genes. The specific genes had similar distribution as substitution/indel/truncation for gene amplification. WD had less specific gene amplification than other HGs, while PD had the highest number of specific genes. WD was enriched in the regulation of fibroblast migration and the negative regulation of transport (Figure S3B); MD was enriched in the positive regulation of cellular processes and the regulation of cell proliferation (Figure S3C); and PD was enriched in the positive regulation of metabolic processes (Figure S3D).

Comparison to the Western population

The findings above were compared against the Western population. An MSKCC dataset from the Western population was downloaded from cBioportal (https://www.cbioportal.org/, accessed on March 5, 2019). With this dataset, nucleotide usage, TMB, driver genes, and biological processes were analyzed using the same procedures as in our dataset. Results showed that WD possessed a higher percentage of transition mutation than PD in the Western population. Meanwhile, PD held a higher percentage of transversion than WD. Such results were in line with those from our dataset. As for the nucleotide usage, only C>G transversion showed higher frequency in PD than in WD (P value =0.017), matching the result from our dataset. Specifically to the Western population, WD had higher A>G mutation than non-WD with P value =0.031. PD had higher A>C and lower A>G substitution than non-PD with P values =0.036 and 7.5e-4, respectively. Among the driver genes, only TP53 mutation was consistently associated with higher HG (P value =1.3e-3, Mann-Whitney U test). Additionally in the Western population, the RB1 mutation tended to be enriched in the high-grade samples.

Further investigation of the similarity between the functional biases for WD- and PD-specific genes revealed an extraordinary consistency. The top significantly enriched biological processes in our dataset showed similar significance in the MSKCC dataset (Figure 5A,B,C). For example, for both our dataset and the MSKCC dataset, PD-specific genes took part in cell proliferation, protein phosphorylation, and regulation of cell proliferation; MD-specific genes took part in the cellular protein modification process and cellular response to stimulus; and WD-specific genes taking part in the regulation of developmental processes and cell differentiation.

Figure 5 Biological processes could predict survival accurately. (A) The top enriched biological process in WD-specific genes from the Chinese population was validated in the Western population; (B) the top enriched biological process in MD-specific genes from the Chinese population was validated in the Western population; (C) the top enriched biological process in PD-specific genes from the Chinese population was validated in the Western population; (D) the intersection of enriched biological processed in the Chinese population and the Western population. WD, well-differentiated; MD, moderately differentiated; PD, poorly differentiated.

It was noteworthy that HG-specific genes may share common enriched biological processes (Figure 5D). To extract the consistent HG-specific biological processes between the two datasets, we first extracted the HG-specific biological processes taken by HG-specific genes for both datasets. Then an intersection was conducted between HG-specific biological processes for both datasets. Through these means, we identified the HG-specific biological processes commonly taken by both datasets. There were 3 WD-specific, 150 MD-specific and 64 PD-specific common biological processes (Table S2). These gene lists were applied for gene ontology enrichment analysis. The PD-specific common biological processes included angiogenesis, phosphatidylinositol-3-phosphate biosynthetic process, glycerophospholipid metabolic process and development of primary male sexual characteristics; the MD-specific common biological processes included response to hydrogen peroxide, response to peptide hormone and protein localization to the nucleus; and the three WD-specific common biological processes were regulation of epithelial to mesenchymal transition involved in endocardial cushion formations, epithelial to mesenchymal transition involved in endocardial cushion formations and regulation of homotypic cell-cell adhesion.


Discussion

Although there have been many studies on the association between gene expression and HG, information on the association between genomic variation and HG is still scarce. The intention of this study was to understand the association between genomic variation and HG and explore the potential of genomic variation as an indicator of HG.

A stable genomic variation pattern should be associated with a hidden molecular mechanism. For example, C>T and C>G substitution could come from DNA editing catalyzed by apolipoprotein B mRNA catalytic subunit-like (APOBEC) and activation-induced deaminase (AID) family, which can bind to both RNA and single-stranded (ss) DNA. DNA deamination by these proteins results in the C>U conversion in single-stranded DNA. Such mutations could result in C>T transition and C>G transversion by different DNA repair polymerases (27). In lung cancer, different cancer subtypes also showed a large difference in C>T transition and C>G transversion (28). Due to the existence of such molecular mechanisms, those stable genomic variation patterns could be stable predictors of HG. In this study, we have analyzed the association of HGs with genomic variation and mutational frequency in the Chinese population and the Western population, and have found a higher C>G transversion mutated in patients with PD HCC for both populations. This association was meaningful in the treatment of such a subset of HCC patients. As reported, APOBEC-related mutagenesis was found to be highly correlated with immunotherapy response (29). Thus, detected C>G transversion could be a good indicator of immunotherapy efficacy. In spite of high C>G transversion being found in HCC and believed as an etiology of HCC by Morishita et al. (30), they did not associate it with any biological significance. Our results revealed that patients with high C>G transversion were strongly associated with poorly differentiated HCC, involving in APOBEC-related mutagenesis.

Taking into account the important mutational scores in relation to survival, we also studied TMB, MSAF, MATH and CMB score, among which only MSAF is significantly associated with HG. As a measure of cellular tumor prevalence, MSAF has been used in many studies (24,31,32). Studies have shown that MSAF is also correlated with tumor burden (31) and several other research studies have revealed that tumor burden is strongly associated with HG (32). Therefore, it is reasonable that MSAF was significantly associated with HG.

Among the driver genes of HCC, TP53 mutation was a consistent biomarker of high HG in both populations, which agreed with the previous studies in ovarian cancer (5) and HCC (33). However, we also noticed difference between the Chinese and Western populations. In the Chinese population, mutations in CTNNB1, ARID2, and ACVR2A were associated with a lower HG, and in the western population, RB1 was associated with a high HG.

During the analysis of biological processes of substitution/indel/truncation and amplification for WD- and PD-specific genes, we found that the biological processes were highly matched for mutation and amplification. For example, WD tumors showed higher substitution/indel/truncation and amplification in the cell differentiation, and PD tumor showed higher substitution/indel/truncation and amplification in the protein phosphorylation. These results demonstrated that a tumor could become WD or PD either through mutations or by amplification, or both. Comparisons between the Chinese and the Western populations also proved that the WD was most enriched in cell differentiation, and the PD was most enriched in phosphorylation. Furthermore, there were also genes for WD or PD involved in phosphorylation or cell differentiation, respectively. A further intersection of their biological processes disclosed the unique biological processes for different HGs. Although these biological processes have been well recognized in basic cancer research, they have not been systematically associated with genomic variations and HG in HCC before.

It should be noted that, instead of ctDNA (circulating tumor DNA) from blood, DNA from the solid tumor was extracted to detect gene mutations. Considering the instability of ctDNA detection, this should be a very important step for applying genomic variations as a predictor of HG. For example, ctDNA is easier to be detected in late-stage cancer and its concentration can be changed by many factors including clinical therapy and tumor development. How the instability of ctDNA detection affects its prediction is another issue to be discussed. The other limitation of this study is that the comparison with the Western population did not include CNV due to the missing information in the MSKCC dataset.

In summary, this pilot study has revealed multiple factors associated with HG. These findings improved our understanding of the molecular mechanism in different HGs of HCC. Further research using ctDNA to detect the genomic variation should be performed to verify this study.

Figure S1 Workflow for this study.
Figure S2 The gene list of the targeted sequencing.
Figure S3 Gene amplification difference between different grades. (A) The gene amplification overlaps between three HGs for substitution/indel/truncation; (B) the enriched biological processes for well differentiated tumors; (C) the enriched biological processes for moderately differentiated tumors; (D) the enriched biological processes for poorly differentiated tumors. The length of the blue bar indicates the negative log transformed false discover rate (FDR). WD, well differentiated; MD, moderately differentiated; PD, poorly differentiated.

Table S1

Clinical characteristics in each histological grade

Characteristics Histological grade
Poor/moderate Well
Age (mean ± SD) 54.9±15.0 60.7±12.5
Gender
   Male 358 44
   Female 54 3
Stage
   I/II/III 375 43
   IV 37 4
Drink
   Yes 50 7
   No 362 40
Family
   Yes 92 5
   No 320 42

Table S2

The HG-specific biological processes

GOID P values_adjusted go_terms Dataset HG-specific
GO:0001525 1.264E-06 Angiogenesis ZB PD
GO:0036092 1.584E-05 Phosphatidylinositol-3-phosphate biosynthetic process MSKCC PD
GO:0006661 9.918E-05 Phosphatidylinositol biosynthetic process MSKCC PD
GO:0006650 0.0002442 Glycerophospholipid metabolic process ZB PD
GO:0036092 0.000248 Phosphatidylinositol-3-phosphate biosynthetic process ZB PD
GO:1902751 0.0002485 Positive regulation of cell cycle G2/M phase transition ZB PD
GO:0006650 0.0003463 Glycerophospholipid metabolic process MSKCC PD
GO:0006661 0.0007599 Phosphatidylinositol biosynthetic process ZB PD
GO:0006644 0.000767 Phospholipid metabolic process ZB PD
GO:0008584 0.0008151 Male gonad development MSKCC PD
GO:0046546 0.0008151 Development of primary male sexual characteristics MSKCC PD
GO:0006644 0.0010525 Phospholipid metabolic process MSKCC PD
GO:0046474 0.0010952 Glycerophospholipid biosynthetic process MSKCC PD
GO:0090218 0.0015026 Positive regulation of lipid kinase activity MSKCC PD
GO:0045017 0.0018219 Glycerolipid biosynthetic process MSKCC PD
GO:0019637 0.0019276 Organophosphate metabolic process ZB PD
GO:0007126 0.002289 Meiotic nuclear division ZB PD
GO:0008654 0.0023611 Phospholipid biosynthetic process MSKCC PD
GO:0043551 0.0026075 Regulation of phosphatidylinositol 3-kinase activity MSKCC PD
GO:1903046 0.0027839 Meiotic cell cycle process ZB PD
GO:1903727 0.0027864 Positive regulation of phospholipid metabolic process MSKCC PD
GO:0001525 0.0030352 Angiogenesis MSKCC PD
GO:0010518 0.0032304 Positive regulation of phospholipase activity ZB PD
GO:0030855 0.0040562 Epithelial cell differentiation MSKCC PD
GO:0035272 0.0042767 Exocrine system development MSKCC PD
GO:0043550 0.0042767 Regulation of lipid kinase activity MSKCC PD
GO:1902749 0.0046541 Regulation of cell cycle G2/M phase transition MSKCC PD
GO:0048146 0.0049414 Positive regulation of fibroblast proliferation MSKCC PD
GO:0033008 0.0050369 Positive regulation of mast cell activation involved in immune response ZB PD
GO:0043306 0.0050369 Positive regulation of mast cell degranulation ZB PD
GO:0046474 0.0050474 Glycerophospholipid biosynthetic process ZB PD
GO:0051321 0.0051928 Meiotic cell cycle ZB PD
GO:0043269 0.0052714 Regulation of ion transport MSKCC PD
GO:0014068 0.0059387 Positive regulation of phosphatidylinositol 3-kinase signaling ZB PD
GO:0033008 0.0062257 Positive regulation of mast cell activation involved in immune response MSKCC PD
GO:0043306 0.0062257 Positive regulation of mast cell degranulation MSKCC PD
GO:0033005 0.0066599 Positive regulation of mast cell activation ZB PD
GO:0045017 0.0071944 Glycerolipid biosynthetic process ZB PD
GO:0044839 0.0072853 Cell cycle G2/M phase transition MSKCC PD
GO:0014068 0.0073914 Positive regulation of phosphatidylinositol 3-kinase signaling MSKCC PD
GO:0002888 0.0080025 Positive regulation of myeloid leukocyte mediated immunity ZB PD
GO:0043302 0.0080025 Positive regulation of leukocyte degranulation ZB PD
GO:0033005 0.0082744 Positive regulation of mast cell activation MSKCC PD
GO:0008654 0.0087335 Phospholipid biosynthetic process ZB PD
GO:0034109 0.0088759 Homotypic cell-cell adhesion ZB PD
GO:0030855 0.0089828 Epithelial cell differentiation ZB PD
GO:1902751 0.0091779 Positive regulation of cell cycle G2/M phase transition MSKCC PD
GO:0006629 0.0093471 Lipid metabolic process MSKCC PD
GO:0008610 0.0100358 Lipid biosynthetic process MSKCC PD
GO:0002888 0.0100358 Positive regulation of myeloid leukocyte mediated immunity MSKCC PD
GO:0043302 0.0100358 Positive regulation of leukocyte degranulation MSKCC PD
GO:0034109 0.0114927 Homotypic cell-cell adhesion MSKCC PD
GO:0051656 0.0119185 Establishment of organelle localization ZB PD
GO:0043269 0.0125078 Regulation of ion transport ZB PD
GO:0060735 0.0129404 Regulation of eif2 alpha phosphorylation by dsRNA ZB PD
GO:0042102 0.0130323 Positive regulation of T cell proliferation ZB PD
GO:0030178 0.0141809 Negative regulation of Wnt signaling pathway ZB PD
GO:0006629 0.0150464 Lipid metabolic process ZB PD
GO:0051656 0.0165508 Establishment of organelle localization MSKCC PD
GO:0060735 0.0165508 Regulation of eif2 alpha phosphorylation by dsRNA MSKCC PD
GO:0007126 0.0170219 Meiotic nuclear division MSKCC PD
GO:0042102 0.0174305 Positive regulation of T cell proliferation MSKCC PD
GO:0090218 0.0181076 Positive regulation of lipid kinase activity ZB PD
GO:0032885 0.0187746 Regulation of polysaccharide biosynthetic process ZB PD
GO:0030178 0.0195602 Negative regulation of Wnt signaling pathway MSKCC PD
GO:1903046 0.0198476 Meiotic cell cycle process MSKCC PD
GO:0008610 0.020913 Lipid biosynthetic process ZB PD
GO:0032752 0.0213557 Positive regulation of interleukin-3 production ZB PD
GO:0042223 0.0213557 Interleukin-3 biosynthetic process ZB PD
GO:0043366 0.0213557 Beta selection ZB PD
GO:0045399 0.0213557 Regulation of interleukin-3 biosynthetic process ZB PD
GO:0045401 0.0213557 Positive regulation of interleukin-3 biosynthetic process ZB PD
GO:0007257 0.0216117 Activation of JUN kinase activity ZB PD
GO:0032881 0.0216117 Regulation of polysaccharide metabolic process ZB PD
GO:1902749 0.0218429 Regulation of cell cycle G2/M phase transition ZB PD
GO:0044839 0.0237717 Cell cycle G2/M phase transition ZB PD
GO:0033003 0.0243556 Regulation of mast cell activation ZB PD
GO:0043551 0.0243556 Regulation of phosphatidylinositol 3-kinase activity ZB PD
GO:1903727 0.0252945 Positive regulation of phospholipid metabolic process ZB PD
GO:0032885 0.0253146 Regulation of polysaccharide biosynthetic process MSKCC PD
GO:0009409 0.0262411 Response to cold ZB PD
GO:1903307 0.0262411 Positive regulation of regulated secretory pathway ZB PD
GO:0032752 0.0274151 Positive regulation of interleukin-3 production MSKCC PD
GO:0042223 0.0274151 Interleukin-3 biosynthetic process MSKCC PD
GO:0043366 0.0274151 Beta selection MSKCC PD
GO:0045399 0.0274151 Regulation of interleukin-3 biosynthetic process MSKCC PD
GO:0045401 0.0274151 Positive regulation of interleukin-3 biosynthetic process MSKCC PD
GO:0008584 0.0282742 Male gonad development ZB PD
GO:0046546 0.0282742 Development of primary male sexual characteristics ZB PD
GO:0002351 0.0282742 Serotonin production involved in inflammatory response ZB PD
GO:0002442 0.0282742 Serotonin secretion involved in inflammatory response ZB PD
GO:0002554 0.0282742 Serotonin secretion by platelet ZB PD
GO:0032252 0.0282742 Secretory granule localization ZB PD
GO:0032672 0.0282742 Regulation of interleukin-3 production ZB PD
GO:0045425 0.0282742 Positive regulation of granulocyte macrophage colony-stimulating factor biosynthetic process ZB PD
GO:0045588 0.0282742 Positive regulation of gamma-delta T cell differentiation ZB PD
GO:1901843 0.0282742 Positive regulation of high voltage-gated calcium channel activity ZB PD
GO:0007257 0.0285333 Activation of JUN kinase activity MSKCC PD
GO:0032881 0.0285333 Regulation of polysaccharide metabolic process MSKCC PD
GO:0042129 0.0307477 Regulation of T cell proliferation ZB PD
GO:0051321 0.0309104 Meiotic cell cycle MSKCC PD
GO:0035272 0.0318221 Exocrine system development ZB PD
GO:0043550 0.0318221 Regulation of lipid kinase activity ZB PD
GO:0033003 0.0322116 Regulation of mast cell activation MSKCC PD
GO:0048146 0.0343113 Positive regulation of fibroblast proliferation ZB PD
GO:0010897 0.0343113 Negative regulation of triglyceride catabolic process ZB PD
GO:0032632 0.0343113 Interleukin-3 production ZB PD
GO:0045423 0.0343113 Regulation of granulocyte macrophage colony-stimulating factor biosynthetic process ZB PD
GO:0060699 0.0343113 Regulation of endoribonuclease activity ZB PD
GO:0007405 0.0343407 Neuroblast proliferation ZB PD
GO:0009409 0.0344727 Response to cold MSKCC PD
GO:1903307 0.0344727 Positive regulation of regulated secretory pathway MSKCC PD
GO:0002351 0.0359801 Serotonin production involved in inflammatory response MSKCC PD
GO:0002442 0.0359801 Serotonin secretion involved in inflammatory response MSKCC PD
GO:0002554 0.0359801 Serotonin secretion by platelet MSKCC PD
GO:0032252 0.0359801 Secretory granule localization MSKCC PD
GO:0032672 0.0359801 Regulation of interleukin-3 production MSKCC PD
GO:0045425 0.0359801 Positive regulation of granulocyte macrophage colony-stimulating factor biosynthetic process MSKCC PD
GO:0045588 0.0359801 Positive regulation of gamma-delta T cell differentiation MSKCC PD
GO:1901843 0.0359801 Positive regulation of high voltage-gated calcium channel activity MSKCC PD
GO:0010518 0.040339 Positive regulation of phospholipase activity MSKCC PD
GO:0042129 0.0411271 Regulation of T cell proliferation MSKCC PD
GO:0010897 0.0437818 Negative regulation of triglyceride catabolic process MSKCC PD
GO:0032632 0.0437818 Interleukin-3 production MSKCC PD
GO:0045423 0.0437818 Regulation of granulocyte macrophage colony-stimulating factor biosynthetic process MSKCC PD
GO:0060699 0.0437818 Regulation of endoribonuclease activity MSKCC PD
GO:0007405 0.0450633 Neuroblast proliferation MSKCC PD
GO:0019637 0.045281 Organophosphate metabolic process MSKCC PD
GO:0042542 1.365E-07 Response to hydrogen peroxide MSKCC MD
GO:0014812 1.111E-06 Muscle cell migration MSKCC MD
GO:0042493 1.147E-06 Response to drug MSKCC MD
GO:1901652 1.569E-06 Response to peptide ZB MD
GO:0044092 1.981E-06 Negative regulation of molecular function ZB MD
GO:0043434 5.246E-06 Response to peptide hormone ZB MD
GO:0000302 1.172E-05 Response to reactive oxygen species MSKCC MD
GO:0034504 1.315E-05 Protein localization to nucleus MSKCC MD
GO:0033365 1.481E-05 Protein localization to organelle MSKCC MD
GO:0070301 2.532E-05 Cellular response to hydrogen peroxide MSKCC MD
GO:0009607 3.12E-05 Response to biotic stimulus MSKCC MD
GO:0034614 4.066E-05 Cellular response to reactive oxygen species MSKCC MD
GO:1904705 4.887E-05 Regulation of vascular smooth muscle cell proliferation MSKCC MD
GO:1990874 4.887E-05 Vascular smooth muscle cell proliferation MSKCC MD
GO:0003279 6.629E-05 Cardiac septum development MSKCC MD
GO:0051707 6.996E-05 Response to other organism MSKCC MD
GO:0043207 7.068E-05 Response to external biotic stimulus MSKCC MD
GO:1901652 7.61E-05 Response to peptide MSKCC MD
GO:0051223 9.568E-05 Regulation of protein transport MSKCC MD
GO:0060411 0.0001143 Cardiac septum morphogenesis MSKCC MD
GO:0014909 0.0001298 Smooth muscle cell migration MSKCC MD
GO:0032496 0.0001321 Response to lipopolysaccharide MSKCC MD
GO:0045682 0.0001383 Regulation of epidermis development MSKCC MD
GO:0006273 0.0001448 Lagging strand elongation MSKCC MD
GO:0043434 0.0001755 Response to peptide hormone MSKCC MD
GO:0002237 0.0001807 Response to molecule of bacterial origin MSKCC MD
GO:0003179 0.000188 Heart valve morphogenesis MSKCC MD
GO:0050864 0.0002231 Regulation of B cell activation MSKCC MD
GO:0003170 0.0002759 Heart valve development MSKCC MD
GO:0051570 0.0003745 Regulation of histone H3-K9 methylation ZB MD
GO:0044092 0.0004357 Negative regulation of molecular function MSKCC MD
GO:0042100 0.0004756 B cell proliferation MSKCC MD
GO:0000302 0.0005009 Response to reactive oxygen species ZB MD
GO:1904019 0.0005234 Epithelial cell apoptotic process MSKCC MD
GO:0006266 0.0005342 DNA ligation ZB MD
GO:0034614 0.00056 Cellular response to reactive oxygen species ZB MD
GO:2001242 0.0005685 Regulation of intrinsic apoptotic signaling pathway MSKCC MD
GO:0046879 0.0006353 Hormone secretion MSKCC MD
GO:0006271 0.0006777 DNA strand elongation involved in DNA replication MSKCC MD
GO:0003205 0.0007089 Cardiac chamber development MSKCC MD
GO:0003007 0.0007878 Heart morphogenesis MSKCC MD
GO:0009914 0.0008096 Hormone transport MSKCC MD
GO:0034504 0.0008947 Protein localization to nucleus ZB MD
GO:0051567 0.000944 Histone H3-K9 methylation ZB MD
GO:0030888 0.001 Regulation of B cell proliferation MSKCC MD
GO:0032611 0.001 Interleukin-1 beta production MSKCC MD
GO:0033157 0.001093 Regulation of intracellular protein transport ZB MD
GO:1902042 0.001286 Negative regulation of extrinsic apoptotic signaling pathway via death domain receptors ZB MD
GO:0050852 0.0012901 T cell receptor signaling pathway MSKCC MD
GO:0006266 0.0012907 DNA ligation MSKCC MD
GO:0032845 0.0013576 Negative regulation of homeostatic process MSKCC MD
GO:0003281 0.0014821 Ventricular septum development MSKCC MD
GO:0033143 0.0014821 Regulation of intracellular steroid hormone receptor signaling pathway MSKCC MD
GO:0022616 0.001571 DNA strand elongation MSKCC MD
GO:0071887 0.0016635 Leukocyte apoptotic process ZB MD
GO:1904035 0.0017433 Regulation of epithelial cell apoptotic process MSKCC MD
GO:0032612 0.0019145 Interleukin-1 production MSKCC MD
GO:0097306 0.0019363 Cellular response to alcohol ZB MD
GO:0034968 0.0021658 Histone lysine methylation ZB MD
GO:0061647 0.002279 Histone H3-K9 modification ZB MD
GO:0033146 0.002285 Regulation of intracellular estrogen receptor signaling pathway MSKCC MD
GO:0022408 0.0023527 Negative regulation of cell-cell adhesion MSKCC MD
GO:0033157 0.0023985 Regulation of intracellular protein transport MSKCC MD
GO:0051223 0.0024086 Regulation of protein transport ZB MD
GO:0032446 0.0024086 Protein modification by small protein conjugation ZB MD
GO:0051573 0.0028681 Negative regulation of histone H3-K9 methylation ZB MD
GO:0018022 0.0028725 Peptidyl-lysine methylation ZB MD
GO:0046824 0.0028725 Positive regulation of nucleocytoplasmic transport ZB MD
GO:0031648 0.0030756 Protein destabilization MSKCC MD
GO:0009636 0.0032692 Response to toxic substance ZB MD
GO:0006261 0.0033055 DNA-dependent DNA replication MSKCC MD
GO:0006273 0.0033374 Lagging strand elongation ZB MD
GO:0002223 0.0035118 Stimulatory C-type lectin receptor signaling pathway ZB MD
GO:0002220 0.0036986 Innate immune response activating cell surface receptor signaling pathway ZB MD
GO:0051103 0.0038896 DNA ligation involved in DNA repair ZB MD
GO:0003007 0.0040602 Heart morphogenesis ZB MD
GO:0016571 0.0042866 Histone methylation ZB MD
GO:0097306 0.0044943 Cellular response to alcohol MSKCC MD
GO:0031060 0.0045733 Regulation of histone methylation ZB MD
GO:0023061 0.0047426 Signal release MSKCC MD
GO:0051573 0.0047426 Negative regulation of histone H3-K9 methylation MSKCC MD
GO:0001666 0.0047462 Response to hypoxia MSKCC MD
GO:0006284 0.0050431 Base-excision repair MSKCC MD
GO:0018205 0.0051017 Peptidyl-lysine modification ZB MD
GO:0036293 0.0053204 Response to decreased oxygen levels MSKCC MD
GO:0061647 0.0053204 Histone H3-K9 modification MSKCC MD
GO:0006261 0.0055276 DNA-dependent DNA replication ZB MD
GO:2001020 0.0055567 Regulation of response to DNA damage stimulus MSKCC MD
GO:0032446 0.0057176 Protein modification by small protein conjugation MSKCC MD
GO:0014909 0.0058867 Smooth muscle cell migration ZB MD
GO:0070664 0.0058867 Negative regulation of leukocyte proliferation ZB MD
GO:0090316 0.0059508 Positive regulation of intracellular protein transport MSKCC MD
GO:0032651 0.0061433 Regulation of interleukin-1 beta production MSKCC MD
GO:0006304 0.0062479 DNA modification MSKCC MD
GO:0051103 0.0062941 DNA ligation involved in DNA repair MSKCC MD
GO:0030520 0.0064205 Intracellular estrogen receptor signaling pathway MSKCC MD
GO:0042093 0.0064205 T-helper cell differentiation MSKCC MD
GO:0045936 0.0065605 Negative regulation of phosphate metabolic process ZB MD
GO:0010563 0.0066057 Negative regulation of phosphorus metabolic process ZB MD
GO:0045604 0.0067768 Regulation of epidermal cell differentiation MSKCC MD
GO:0031061 0.0067822 Negative regulation of histone methylation ZB MD
GO:0034968 0.0067993 Histone lysine methylation MSKCC MD
GO:0002294 0.0071277 CD4-positive, alpha-beta T cell differentiation involved in immune response MSKCC MD
GO:0002287 0.0074349 alpha-beta T cell activation involved in immune response MSKCC MD
GO:0002293 0.0074349 alpha-beta T cell differentiation involved in immune response MSKCC MD
GO:0031663 0.0074349 Lipopolysaccharide-mediated signaling pathway MSKCC MD
GO:0038034 0.0077505 Signal transduction in absence of ligand ZB MD
GO:0097192 0.0077505 Extrinsic apoptotic signaling pathway in absence of ligand ZB MD
GO:0034284 0.0078981 Response to monosaccharide MSKCC MD
GO:0006271 0.0081662 DNA strand elongation involved in DNA replication ZB MD
GO:0046660 0.0082366 Female sex differentiation MSKCC MD
GO:0014812 0.008241 Muscle cell migration ZB MD
GO:0003283 0.0088126 Atrial septum development ZB MD
GO:0023019 0.0088126 Signal transduction involved in regulation of gene expression ZB MD
GO:0007259 0.0089793 JAK-STAT cascade ZB MD
GO:0097696 0.0089793 STAT cascade ZB MD
GO:0018022 0.0091089 Peptidyl-lysine methylation MSKCC MD
GO:0046824 0.0091089 Positive regulation of nucleocytoplasmic transport MSKCC MD
GO:0001541 0.0092698 Ovarian follicle development MSKCC MD
GO:0070301 0.0095041 Cellular response to hydrogen peroxide ZB MD
GO:0002292 0.0096977 T cell differentiation involved in immune response MSKCC MD
GO:1903533 0.0097929 Regulation of protein targeting ZB MD
GO:0006479 0.0099192 Protein methylation ZB MD
GO:0008213 0.0099192 Protein alkylation ZB MD
GO:0050852 0.0099192 T cell receptor signaling pathway ZB MD
GO:0032845 0.0101971 Negative regulation of homeostatic process ZB MD
GO:0031060 0.0104978 Regulation of histone methylation MSKCC MD
GO:0032652 0.0104978 Regulation of interleukin-1 production MSKCC MD
GO:0008625 0.0108141 Extrinsic apoptotic signaling pathway via death domain receptors ZB MD
GO:0032496 0.0108141 Response to lipopolysaccharide ZB MD
GO:0009743 0.0108748 Response to carbohydrate MSKCC MD
GO:0001947 0.0108748 Heart looping MSKCC MD
GO:0048678 0.0108748 Response to axon injury MSKCC MD
GO:0072091 0.0108748 Regulation of stem cell proliferation MSKCC MD
GO:1903533 0.0110206 Regulation of protein targeting MSKCC MD
GO:0002223 0.0110206 Stimulatory C-type lectin receptor signaling pathway MSKCC MD
GO:0031061 0.0110206 Negative regulation of histone methylation MSKCC MD
GO:0043367 0.0112179 CD4-positive, alpha-beta T cell differentiation MSKCC MD
GO:0002220 0.0115502 Innate immune response activating cell surface receptor signaling pathway MSKCC MD
GO:1904705 0.0117161 Regulation of vascular smooth muscle cell proliferation ZB MD
GO:1990874 0.0117161 Vascular smooth muscle cell proliferation ZB MD
GO:0042100 0.0117593 B cell proliferation ZB MD
GO:1904019 0.0124536 Epithelial cell apoptotic process ZB MD
GO:0006471 0.0125445 Protein ADP-ribosylation ZB MD
GO:0002237 0.0127762 Response to molecule of bacterial origin ZB MD
GO:0061371 0.0130477 Determination of heart left/right asymmetry MSKCC MD
GO:0016571 0.0132027 Histone methylation MSKCC MD
GO:0022616 0.0132769 DNA strand elongation ZB MD
GO:0008585 0.0132769 Female gonad development ZB MD
GO:0003143 0.0134719 Embryonic heart tube morphogenesis MSKCC MD
GO:0008589 0.0134719 Regulation of smoothened signaling pathway MSKCC MD
GO:0035710 0.0134719 CD4-positive, alpha-beta T cell activation MSKCC MD
GO:0070664 0.0134719 Negative regulation of leukocyte proliferation MSKCC MD
GO:0071301 0.0136096 Cellular response to vitamin B1 ZB MD
GO:0090347 0.0136096 Regulation of cellular organohalogen metabolic process ZB MD
GO:0090348 0.0136096 Regulation of cellular organofluorine metabolic process ZB MD
GO:0090349 0.0136096 Negative regulation of cellular organohalogen metabolic process ZB MD
GO:0090350 0.0136096 Negative regulation of cellular organofluorine metabolic process ZB MD
GO:1904404 0.0136096 Response to formaldehyde ZB MD
GO:0003279 0.0136766 Cardiac septum development ZB MD
GO:0046545 0.0139992 Development of primary female sexual characteristics ZB MD
GO:0009743 0.0140538 Response to carbohydrate ZB MD
GO:0043086 0.0143878 Negative regulation of catalytic activity ZB MD
GO:0003283 0.0143938 Atrial septum development MSKCC MD
GO:0023019 0.0143938 Signal transduction involved in regulation of gene expression MSKCC MD
GO:0007389 0.015078 Pattern specification process MSKCC MD
GO:0006304 0.0153019 DNA modification ZB MD
GO:0051100 0.0154442 Negative regulation of binding MSKCC MD
GO:0045936 0.0155204 Negative regulation of phosphate metabolic process MSKCC MD
GO:0051570 0.015525 Regulation of histone H3-K9 methylation MSKCC MD
GO:0010563 0.01554 Negative regulation of phosphorus metabolic process MSKCC MD
GO:0046634 0.0155981 Regulation of alpha-beta T cell activation MSKCC MD
GO:0071301 0.0155981 Cellular response to vitamin B1 MSKCC MD
GO:0090347 0.0155981 Regulation of cellular organohalogen metabolic process MSKCC MD
GO:0090348 0.0155981 Regulation of cellular organofluorine metabolic process MSKCC MD
GO:0090349 0.0155981 Negative regulation of cellular organohalogen metabolic process MSKCC MD
GO:0090350 0.0155981 Negative regulation of cellular organofluorine metabolic process MSKCC MD
GO:1904404 0.0155981 Response to formaldehyde MSKCC MD
GO:0033146 0.0159353 Regulation of intracellular estrogen receptor signaling pathway ZB MD
GO:0051051 0.0167241 Negative regulation of transport MSKCC MD
GO:0038034 0.0167508 Signal transduction in absence of ligand MSKCC MD
GO:0097192 0.0167508 Extrinsic apoptotic signaling pathway in absence of ligand MSKCC MD
GO:0046660 0.0191336 Female sex differentiation ZB MD
GO:0031648 0.01952 Protein destabilization ZB MD
GO:0006471 0.0196997 Protein ADP-ribosylation MSKCC MD
GO:0043086 0.0202833 Negative regulation of catalytic activity MSKCC MD
GO:0003179 0.0203264 Heart valve morphogenesis ZB MD
GO:0003230 0.0203264 Cardiac atrium development ZB MD
GO:0033365 0.0219486 Protein localization to organelle ZB MD
GO:0042542 0.0231473 Response to hydrogen peroxide ZB MD
GO:0010868 0.0231473 Negative regulation of triglyceride biosynthetic process ZB MD
GO:0071460 0.0231473 Cellular response to cell-matrix adhesion ZB MD
GO:0090345 0.0231473 Cellular organohalogen metabolic process ZB MD
GO:0090346 0.0231473 Cellular organofluorine metabolic process ZB MD
GO:0050864 0.0232449 Regulation of B cell activation ZB MD
GO:0044262 0.0234314 Cellular carbohydrate metabolic process MSKCC MD
GO:0003170 0.0234325 Heart valve development ZB MD
GO:0008625 0.0238195 Extrinsic apoptotic signaling pathway via death domain receptors MSKCC MD
GO:0043029 0.0244019 T cell homeostasis ZB MD
GO:0046825 0.0252439 Regulation of protein export from nucleus ZB MD
GO:0018205 0.0254654 Peptidyl-lysine modification MSKCC MD
GO:0090316 0.0255328 Positive regulation of intracellular protein transport ZB MD
GO:0022408 0.0260472 Negative regulation of cell-cell adhesion ZB MD
GO:0006284 0.0260472 Base-excision repair ZB MD
GO:0010883 0.0260472 Regulation of lipid storage ZB MD
GO:0007259 0.0262577 JAK-STAT cascade MSKCC MD
GO:0097696 0.0262577 STAT cascade MSKCC MD
GO:0051567 0.0262577 Histone H3-K9 methylation MSKCC MD
GO:0010868 0.0262577 Negative regulation of triglyceride biosynthetic process MSKCC MD
GO:0071460 0.0262577 Cellular response to cell-matrix adhesion MSKCC MD
GO:0090345 0.0262577 Cellular organohalogen metabolic process MSKCC MD
GO:0090346 0.0262577 Cellular organofluorine metabolic process MSKCC MD
GO:0044262 0.0265122 Cellular carbohydrate metabolic process ZB MD
GO:0042493 0.0271005 Response to drug ZB MD
GO:0006479 0.0275738 Protein methylation MSKCC MD
GO:0008213 0.0275738 Protein alkylation MSKCC MD
GO:0008585 0.0277022 Female gonad development MSKCC MD
GO:0051100 0.029382 Negative regulation of binding ZB MD
GO:0032651 0.0295013 Regulation of interleukin-1 beta production ZB MD
GO:0043525 0.0295013 Positive regulation of neuron apoptotic process ZB MD
GO:0051055 0.0295013 Negative regulation of lipid biosynthetic process ZB MD
GO:0007389 0.0295013 Pattern specification process ZB MD
GO:0023061 0.0295013 Signal release ZB MD
GO:0030520 0.0295013 Intracellular estrogen receptor signaling pathway ZB MD
GO:0042093 0.0295013 T-helper cell differentiation ZB MD
GO:0009822 0.0295013 Alkaloid catabolic process ZB MD
GO:0010266 0.0295013 Response to vitamin B1 ZB MD
GO:0033076 0.0295013 Isoquinoline alkaloid metabolic process ZB MD
GO:0071494 0.0295013 Cellular response to UV-C ZB MD
GO:0098760 0.0295013 Response to interleukin-7 ZB MD
GO:0098761 0.0295013 Cellular response to interleukin-7 ZB MD
GO:1990785 0.0295013 Response to water-immersion restraint stress ZB MD
GO:0071887 0.029714 Leukocyte apoptotic process MSKCC MD
GO:0045604 0.0298719 Regulation of epidermal cell differentiation ZB MD
GO:0046545 0.0305378 Development of primary female sexual characteristics MSKCC MD
GO:1902042 0.0306947 Negative regulation of extrinsic apoptotic signaling pathway via death domain receptors MSKCC MD
GO:0002294 0.0309829 CD4-positive, alpha-beta T cell differentiation involved in immune response ZB MD
GO:0002287 0.0320739 alpha-beta T cell activation involved in immune response ZB MD
GO:0002293 0.0320739 alpha-beta T cell differentiation involved in immune response ZB MD
GO:0031663 0.0320739 Lipopolysaccharide-mediated signaling pathway ZB MD
GO:0003230 0.0321242 Cardiac atrium development MSKCC MD
GO:2001242 0.0323501 Regulation of intrinsic apoptotic signaling pathway ZB MD
GO:0051051 0.0324454 Negative regulation of transport ZB MD
GO:0051707 0.0338559 Response to other organism ZB MD
GO:0043207 0.033997 Response to external biotic stimulus ZB MD
GO:0009822 0.0354738 Alkaloid catabolic process MSKCC MD
GO:0010266 0.0354738 Response to vitamin B1 MSKCC MD
GO:0033076 0.0354738 Isoquinoline alkaloid metabolic process MSKCC MD
GO:0071494 0.0354738 Cellular response to UV-C MSKCC MD
GO:0098760 0.0354738 Response to interleukin-7 MSKCC MD
GO:0098761 0.0354738 Cellular response to interleukin-7 MSKCC MD
GO:1990785 0.0354738 Response to water-immersion restraint stress MSKCC MD
GO:0003205 0.0358613 Cardiac chamber development ZB MD
GO:0001541 0.0363016 Ovarian follicle development ZB MD
GO:0007089 0.0363016 Traversing start control point of mitotic cell cycle ZB MD
GO:0010989 0.0363016 Negative regulation of low-density lipoprotein particle clearance ZB MD
GO:0014042 0.0363016 Positive regulation of neuron maturation ZB MD
GO:0035799 0.0363016 Ureter maturation ZB MD
GO:0042997 0.0363016 Negative regulation of Golgi to plasma membrane protein transport ZB MD
GO:0070427 0.0363016 Nucleotide-binding oligomerization domain containing 1 signaling pathway ZB MD
GO:2000048 0.0363016 Negative regulation of cell-cell adhesion mediated by cadherin ZB MD
GO:0046879 0.0364954 Hormone secretion ZB MD
GO:0002292 0.0371894 T cell differentiation involved in immune response ZB MD
GO:0030888 0.0371894 Regulation of B cell proliferation ZB MD
GO:0032611 0.0371894 Interleukin-1 beta production ZB MD
GO:0043029 0.0386747 T cell homeostasis MSKCC MD
GO:0032652 0.0392552 Regulation of interleukin-1 production ZB MD
GO:2001020 0.039929 Regulation of response to DNA damage stimulus ZB MD
GO:0046825 0.0400926 Regulation of protein export from nucleus MSKCC MD
GO:0001947 0.040316 Heart looping ZB MD
GO:0048678 0.040316 Response to axon injury ZB MD
GO:0072091 0.040316 Regulation of stem cell proliferation ZB MD
GO:0009914 0.0407189 Hormone transport ZB MD
GO:0009607 0.0410816 Response to biotic stimulus ZB MD
GO:0043367 0.0414048 CD4-positive, alpha-beta T cell differentiation ZB MD
GO:0010883 0.0416577 Regulation of lipid storage MSKCC MD
GO:0007089 0.0432135 Traversing start control point of mitotic cell cycle MSKCC MD
GO:0010989 0.0432135 Negative regulation of low-density lipoprotein particle clearance MSKCC MD
GO:0014042 0.0432135 Positive regulation of neuron maturation MSKCC MD
GO:0035799 0.0432135 Ureter maturation MSKCC MD
GO:0042997 0.0432135 Negative regulation of Golgi to plasma membrane protein transport MSKCC MD
GO:0070427 0.0432135 Nucleotide-binding oligomerization domain containing 1 signaling pathway MSKCC MD
GO:2000048 0.0432135 Negative regulation of cell-cell adhesion mediated by cadherin MSKCC MD
GO:0003281 0.0437357 Ventricular septum development ZB MD
GO:0033143 0.0437357 Regulation of intracellular steroid hormone receptor signaling pathway ZB MD
GO:0060411 0.0437357 Cardiac septum morphogenesis ZB MD
GO:0061371 0.0447239 Determination of heart left/right asymmetry ZB MD
GO:0009636 0.0455879 Response to toxic substance MSKCC MD
GO:0003143 0.0456721 Embryonic heart tube morphogenesis ZB MD
GO:0008589 0.0456721 Regulation of smoothened signaling pathway ZB MD
GO:0035710 0.0456721 CD4-positive, alpha-beta T cell activation ZB MD
GO:0001666 0.0462698 Response to hypoxia ZB MD
GO:0043525 0.0463606 Positive regulation of neuron apoptotic process MSKCC MD
GO:0051055 0.0463606 Negative regulation of lipid biosynthetic process MSKCC MD
GO:0045682 0.0465784 Regulation of epidermis development ZB MD
GO:1904035 0.0465784 Regulation of epithelial cell apoptotic process ZB MD
GO:0034284 0.0473956 Response to monosaccharide ZB MD
GO:0032612 0.0473956 Interleukin-1 production ZB MD
GO:0036293 0.047653 Response to decreased oxygen levels ZB MD
GO:0046634 0.0493719 Regulation of alpha-beta T cell activation ZB MD
GO:1905005 0.0303648 Regulation of epithelial to mesenchymal transition involved in endocardial cushion formation ZB WD
GO:0003198 0.0462411 Epithelial to mesenchymal transition involved in endocardial cushion formation ZB WD
GO:0034110 0.0498585 Regulation of homotypic cell-cell adhesion ZB WD
GO:1905005 0.027475 Regulation of epithelial to mesenchymal transition involved in endocardial cushion formation MSKCC WD
GO:0003198 0.0414965 Epithelial to mesenchymal transition involved in endocardial cushion formation MSKCC WD
GO:0034110 0.0441837 Regulation of homotypic cell-cell adhesion MSKCC WD

Note: dataset, our dataset was named ZB. PD, poor differentiated; MD, moderate differentiated; WD, well differentiated.


Acknowledgments

Funding: None.


Footnote

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/tcr.2020.03.32). The work was carried out as part of the employment of the corresponding author at the Affiliated Hospital of Qingdao University. The Affiliated Hospital of Qingdao University was not involved in the manuscript writing, editing, approval, or decision to publish. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. This study was approved by Shandong Provincial Hospital Affiliated to Shandong University and The Affiliated Hospital of Qingdao University. Each participant provided written informed consent. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013).

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Cillo U, Giuliani T, Polacco M, et al. Prediction of hepatocellular carcinoma biological behavior in patient selection for liver transplantation. World J Gastroenterol 2016;22:232-52. [Crossref] [PubMed]
  2. Schadendorf D, Hodi FS, Robert C, et al. Pooled Analysis of Long-Term Survival Data From Phase II and Phase III Trials of Ipilimumab in Unresectable or Metastatic Melanoma. J Clin Oncol 2015;33:1889-94. [Crossref] [PubMed]
  3. Kobayashi M, Sawada K, Nakamura K, et al. Exosomal miR-1290 is a potential biomarker of high-grade serous ovarian carcinoma and can discriminate patients from those with malignancies of other histological types. J Ovarian Res 2018;11:81. [Crossref] [PubMed]
  4. Skoog P, Ohlsson M, Ferno M, et al. Tumor tissue protein signatures reflect histological grade of breast cancer. PLoS One 2017;12:e0179775. [Crossref] [PubMed]
  5. Cole AJ, Dwight T, Gill AJ, et al. Assessing mutant p53 in primary high-grade serous ovarian cancer using immunohistochemistry and massively parallel sequencing. Sci Rep 2016;6:26191. [Crossref] [PubMed]
  6. Deng L, Xiong P, Luo Y, et al. Association between IDH1/2 mutations and brain glioma grade. Oncol Lett 2018;16:5405-9. [PubMed]
  7. Wodziński D, Wosiak A, Pietrzak J, et al. Does the expression of the ACVR2A gene affect the development of colorectal cancer? Genet Mol Biol 2019;42:32-9. [Crossref] [PubMed]
  8. Edmondson HA, Steiner PE. Primary carcinoma of the liver: a study of 100 cases among 48,900 necropsies. Cancer 1954;7:462-503. [Crossref] [PubMed]
  9. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet Journal 2011;17:3. [Crossref]
  10. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 2009;25:1754-60. [Crossref] [PubMed]
  11. McKenna A, Hanna M, Banks E, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 2010;20:1297-303. [Crossref] [PubMed]
  12. Cibulskis K, Lawrence MS, Carter SL, et al. point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 2013;31:213-9. [Crossref] [PubMed]
  13. Koboldt DC, Chen K, Wylie T, et al. VarScan: variant detection in massively parallel sequencing of individual and pooled samples. Bioinformatics 2009;25:2283-5. [Crossref] [PubMed]
  14. Karczewski KJ, Weisburd B, Thomas B, et al. The ExAC browser: displaying reference data information from over 60 000 exomes. Nucleic Acids Res 2017;45:D840-D845. [Crossref] [PubMed]
  15. Lek M, Karczewski KJ, Minikel EV, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 2016;536:285-91. [Crossref] [PubMed]
  16. 1000 Genomes Project Consortium, Abecasis GR, Altshuler D, et al. A map of human genome variation from population-scale sequencing. Nature 2010;467:1061-73.
  17. NHLBI. Exome Variant Server. Available online: https://evs.gs.washington.edu. 2017/7/1.
  18. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res 2010;38:e164. [Crossref] [PubMed]
  19. Li H. FermiKit: assembly-based variant calling for Illumina resequencing data. Bioinformatics 2015;31:3694-6. [PubMed]
  20. Talevich E, Shain AH, Botton T, et al. CNVkit: Genome-Wide Copy Number Detection and Visualization from Targeted DNA Sequencing. PLoS Comput Biol 2016;12:e1004873. [Crossref] [PubMed]
  21. Murtaza M, Dawson SJ, Pogrebniak K, et al. Multifocal clonal evolution characterized using circulating tumour DNA in a case of metastatic breast cancer. Nat Commun 2015;6:8760. [Crossref] [PubMed]
  22. Jiang T, Shi W, Wali VB, et al. Predictors of Chemosensitivity in Triple Negative Breast Cancer: An Integrated Genomic Analysis. PLoS Med 2016;13:e1002193. [Crossref] [PubMed]
  23. Edge SB, Byrd D, Compton C, et al. Digestive system. In: Trotti A. editor. AJCC cancer staging manual, ed 7th. New York: Springer, 2010.
  24. Schrock AB, Pavlick D, Klempner SJ, et al. Hybrid Capture-Based Genomic Profiling of Circulating Tumor DNA from Patients with Advanced Cancers of the Gastrointestinal Tract or Anus. Clin Cancer Res 2018;24:1881-90. [Crossref] [PubMed]
  25. Lee JS. The mutational landscape of hepatocellular carcinoma. Clin Mol Hepatol 2015;21:220-9. [Crossref] [PubMed]
  26. Ding XX, Zhu QG, Zhang SM, et al. Precision medicine for hepatocellular carcinoma: driver mutations and targeted therapy. Oncotarget 2017;8:55715-30. [PubMed]
  27. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature 2013;500:415-21. [Crossref] [PubMed]
  28. Kandoth C, McLellan MD, Vandin F, et al. Mutational landscape and significance across 12 major cancer types. Nature 2013;502:333-9. [Crossref] [PubMed]
  29. Boichard A, Pham TV, Yeerna H, et al. APOBEC-related mutagenesis and neo-peptide hydrophobicity: implications for response to immunotherapy. Oncoimmunology 2018;8:1550341. [Crossref] [PubMed]
  30. Morishita A, Iwama H, Fujihara S, et al. Targeted sequencing of cancer-associated genes in hepatocellular carcinoma using next-generation sequencing. Oncol Lett 2018;15:528-32. [PubMed]
  31. Zhou C, Yuan Z, Ma W, et al. Clinical utility of tumor genomic profiling in patients with high plasma circulating tumor DNA burden or metabolically active tumors. J Hematol Oncol 2018;11:129. [Crossref] [PubMed]
  32. Badowska-Kozakiewicz AM, Liszcz A, Sobol M, et al. Retrospective evaluation of histopathological examinations in invasive ductal breast cancer of no special type: an analysis of 691 patients. Arch Med Sci 2017;13:1408-15. [Crossref] [PubMed]
  33. Kancherla V, Abdullazade S, Matter MS, et al. Genomic Analysis Revealed New Oncogenic Signatures in TP53-Mutant Hepatocellular Carcinoma. Front Genet 2018;9:2. [Crossref] [PubMed]
Cite this article as: Liu J, Li G, Guo Y, Fan N, Zang Y. The association between genomic variations and histological grade in hepatocellular carcinoma. Transl Cancer Res 2020;9(4):2424-2433. doi: 10.21037/tcr.2020.03.32

Download Citation